throbber
Received: 15 December 2020
`
`Revised: 2 June 2021
`
`Accepted: 7 June 2021
`
`DOI: 10.1002/jha2.252
`
`REVIEW ARTICLE
`
`
`
`British Society for
`Haematology
`
`® Checkfor updates
`
`The beginning of a new therapeutic era in acute myeloid
`leukemia
`
`Christian Récher ©
`
`Service d’Hématologie, Centre Hospitalier
`Universitaire de Toulouse,Institut
`Universitaire du Cancer de Toulouse
`Oncopole, Université ToulouseIII Paul
`Sabatier, Centre de Recherches en
`Cancérologie de Toulouse, Toulouse, France
`
`Correspondence
`Christian Récher, Service d’Hématologie,
`Centre Hospitalier Universitaire de Toulouse,
`Institut Universitaire du Cancer de Toulouse
`Oncopole, Université ToulouseII Paul Sabatier,
`Centre de Recherches en Cancérologie de
`Toulouse, Toulouse, France.
`Email: recher.christian@iuct-oncopole.fr
`
`Abstract
`
`In the field of AML, the early 2000s were shaped by the advent of novel molecular
`
`biology technologies including high-throughput sequencing that improved prognostic
`
`classification, response evaluation through the quantification of minimal residual dis-
`
`ease, and the launchof research on targeted therapies. Our knowledge of leukemoge-
`
`nesis, AML genetic diversity, gene-gene interactions, clonal evolution, and treatment
`
`response assessmenthasalso greatly improved. Newclassifications based on chromo-
`
`somal abnormalities and gene mutations are now integrated on a routine basis. These
`
`considerable efforts contributed to the discovery and developmentof promising drugs
`
`whichspecifically target gene mutations, apoptotic pathways andcell surface antigens
`
`as well as reformulate classical cytotoxic agents. In less than 2 years, nine novels drugs
`
`have been approved for the treatment of AML patients, and many others are being
`
`intensively investigated,in particular immune therapies. There are now numerous clini-
`
`calresearch opportunities offered to clinicians, thanks to these new treatment options.
`
`Weareonly at the start of anew era which should see major disruptions in the way we
`
`understand, treat, and monitor patients with AML.
`
`KEYWORDS
`
`acute myeloid leukemia, CPX-351, enasidenib, FLT3 inhibitors, gemtuzumab ozogamycin,gilter-
`itinib, glasdegib, IDH inhibitors, ivosidenib, midostaurin, monoclonal antibodies, oral azacitidine,
`TP53, venetoclax
`
`1
`
`|
`
`INTRODUCTION
`
`It has long been written that there was no breakthrough for the
`treatment of acute myeloid leukemia (AML) as compared to other
`hematological malignancies including chronic myeloid leukemia, B-cell
`lymphoma (BCL), or multiple myeloma [1]. Going back more than
`20 years ago, the situation was roughly similar for multiple myeloma.
`Youngerpatients were treated with high-dose melphalan and autolo-
`gous stem-cell transplantation while older patients received low-dose
`melphalan and prednisone, the so-called MP regimen that has fallen
`
`into oblivion over the years [2]. Now, there have been more than 10
`novel drugs approved in myelomatargeting intracellular pathways,
`tumor microenvironment, and cell surface antigens the combinations
`of which have transformed a highly deadly disease into a chronic
`one[3, 4]. In AML, we have beenrelying for the past 40 years on the
`combination of cytarabine and an anthracycline, so-called (“7+3”), as
`induction chemotherapy in patients suitable for intensive treatments
`followed by high-dose cytarabine consolidation and eventually by
`allogeneic stem cell transplantation (cure being the goal), whereas
`older or unfit patients received low-dose cytarabine or more recently
`
`This is an open accessarticle under the terms of the Creative CommonsAttribution License, which permits use, distribution and reproduction in any medium, provided
`the original work is properly cited.
`© 2021 The Authors. eJHaem published by British Society for Haematology and John Wiley & SonsLtd.
`
`eJHaem. 2021;2:823-833.
`wileyonlinelibrary.com/journal/jha2
`823
`
`CELGENE 2110
`APOTEX v. CELGENE
`IPR2023-00512
`
`

`

`4| WILEY
`
`RECHER
`
`TABLE1=Novel drugs recently approvedin acute myeloid leukemia
`
`Agent
`
`Midostaurin
`
`Enasidenib
`
`CPX-351
`
`Approval
`
`2017
`
`Mechanismofaction
`
`FLT3inhibition
`
`2017
`
`2017
`
`IDH2 mutantinhibition
`
`Liposomal formulation
`including daunorubicin
`and cytarabineat a fixed
`5-molar:1-molar ratio
`
`CD33 monoclonal antibody
`linked to calicheamicin
`
`IDH1 mutantinhibition
`
`Hedgehog pathway
`inhibition
`
`Bcl-2 inhibition
`
`FLT3inhibition
`
`Hypomethylating agent
`
`Indication
`
`Newly diagnosed AML with FLT3 mutation
`in combination with standard induction,
`consolidation +/-maintenance
`
`Relapsed or refractory AML with IDH2
`mutation
`
`Newly diagnosed, therapy-related AML
`(t-AML) or AML with
`myelodysplasia-related
`
`Newly diagnosed, CD33-positive AML in
`combination with standard induction and
`relapsed or refractory CD33-positive
`AML
`
`Relapsed or refractory or newly diagnosed
`(unfit) AML with IDH1 mutation
`
`Unfit or older (>75y) patients with newly
`diagnosed AML in combination with
`low-dose cytarabine
`
`Unfit or older (>75y) patients with newly
`diagnosed AML in combination with
`hypomethylating agents or low-dose
`cytarabine
`
`Relapsed or refractory AML with FLT3
`mutation
`
`Continued treatment of AML patients who
`achieved CR/CRifollowing intensive
`induction chemotherapyand are not able
`to complete intensive curative therapy
`(i.e., alloSCT)
`
`changes (AML-MRC)
`
`Gemtuzumab ozogamicin
`
`2017
`
`lvosidenib
`
`Glasdegib
`
`Venetoclax
`
`Gilteritinib
`
`Oral azacitidine
`
`2018
`
`2018
`
`2018
`
`2018
`
`2020
`
`hypomethylating agents, both inducing few complete responses (CRs)
`andlittle hope for cure (prolonged survival being the goal) [5]. Then,
`genomics changed the game.Firstly, targeted gene sequencing succes-
`sively identified FLT3-ITD, CEBPA, and NPM1 mutations which helped
`improve prognostic classification, the response evaluation through
`quantification of molecular residual disease, and launched the research
`on targeted therapies leading to the registration of midostaurin as the
`first FLT3 inhibitor in AML [6, 7]. Soon after, thanks to the advance
`in high-throughput sequencing technologies, the first AML genome
`was reported in 2008 and subsequent studies identified several
`novel recurrent mutations with pathophysiological, prognostic, or
`therapeutic relevance such as |DH1, IDH2, or TP53 mutations [8-10].
`Our knowledge of leukemogenesis, AML genetic diversity, gene-gene
`interactions, clonal evolution, and treatment response assessmenthas
`also greatly improved, and new classifications based on chromosomal
`abnormalities and gene mutations are now integrated in clinical
`environments [11-17]. These considerable efforts contributed to the
`discovery and development of promising drugs for AML specifically
`targeting gene mutations, apoptotic pathways, and cell surface anti-
`gens. Novel liposomal formulations of classical cytotoxic agents are
`also promising. Midostaurin, gemtuzumab ozogamycin, glasdegib,
`
`venetoclax, ivosidenib, enasidenib,gilteritinib, CPX-351, and oral azac-
`itidine were approved by the Food and Drug Administration (FDA) of
`for AML patientsin less than 3 years between 2017 and 2020[18, 19]
`(Table 1).
`Rather than just another review stacking up one novel drug after
`another, this article will try to outline the perspectives for the com-
`ing years in the field of AML [20, 21]. Indeed, there are now numer-
`ous clinical research opportunities offered to clinicians with these
`new molecules and with those under development. With this sudden
`over-abundanceofchoices, wefeel like a lottery player who gets rich
`overnight and wonders whathe’s going to do withitall!
`
`2 | VENETOCLAX IN AML, THE MAGIC
`POTENTIATOR
`
`The anti-apoptotic BCL 2 (BCL-2) protein is overexpressed in AML,
`especially in leukemic stem cells that are supposed to be responsible
`for chemoresistance and relapse. BCL-2 overexpression is a poor
`risk factor in AML and is associated with chemoresistance [22].
`BCL-2 inhibition by small molecule inhibitors kills AML blasts, targets
`
`

`

`RECHER
`
`WILEY
`
`oxidative phosphorylation, and selectively eradicates leukemic stem
`cells [23, 24].
`In AML patients, contrary to chronic lymphocytic
`leukemia, venetoclax - an oral, selective, small-molecule inhibitor of
`
`comparable, and toxicity is likely to be lower with the doublet, although
`this important question should be the matter of prospective random-
`ized trials.
`
`Beyond the population of unfit patients, venetoclax is likely to
`become the drug of choice for combination therapy in virtually all
`AML patients. In relapse or refractory AML, the preliminary results
`of combinations with other small molecules inhibitors such as FLT3,
`
`IDH, or MDM2 inhibitors seem very promising [37, 38]. In patientsfit
`for intensive chemotherapy, adding venetoclax to 7+3 is feasible and
`induces promising responserates, especially in patients with interme-
`diate cytogenetic risk or with NPM1 or IDH mutations [25]. In these fit
`patients who have achieved CR after intensive induction chemother-
`apy, venetoclax in combination with intermediate dose cytarabine as
`consolidation therapy and/or with azacitidine as maintenance therapy
`so as to eradicate residual disease will be an important scope of inves-
`tigation. To date, 91 clinical trials with venetoclax in AML are recorded
`on theclinicaltrials.gov Website, thus highlighting the tremendous
`interest for this drug in AML. This has been confirmed at the virtual
`2020 American Society of Hematology meeting where noless than 10
`oral communications dealt with venetoclax based-combinations. Vene-
`
`toclax is becoming the drug of choice to combinewithall kind of ther-
`apeutic strategies in AML including high or low intensity chemother-
`apy, novel agents, and tyrosine kinase inhibitors. In a phase 2 trial, a
`low-intensity backboneof cladribine/low dose cytarabine plus veneto-
`clax alternating with azacitidine plus venetoclax for older patients with
`AML,yielded very high rates of durable MRD negative CR (96% CR/CRi
`and 80% MRD negative). With a median follow-up of 11+ months, the
`median OS wasnotreached, with 6- and 12-month OSrates of 86%
`
`and 70%, respectively [39]. Venetoclax has been also combined with
`intensive chemotherapyincluding CPX-351 or FLAG-Ida regimen [40,
`41]. These combinationsare feasible provided that the dose and dura-
`tion of venetoclax exposure is reduced. Prolonged myelosuppression
`and infections remain a major problem with this type of combinations
`which will be reserved for suitable patients. In FLT3 mutated AML,
`combining venetoclax plus FLT3 inhibitors such as quizartinib orgilter-
`itinib with or without hypomethylating agents also induced impressive
`responserates in R/R AML patients [42, 43]. However, AML patients
`with TP53 mutations did not appear to benefit from venetoclax based-
`treatment. Indeed, the combination of venetoclax and decitabine was
`
`associated with inferior response rate, shorter response duration,
`higher MRDpositivity, and a poor median OSof 5.2 months[44].
`
`BCL-2 - is not very active as a single agent with perhaps, the excep-
`tion of NPM1 or IDH mutation subgroups [25, 26]. However, when
`combined to low-dose cytarabine or with hypomethylating agents,
`venetoclax dramatically increases the CR rates, thus demonstrating
`synergistic activity in patients. Venetoclax has been recently approved
`in combination with hypomethylating agents asfirst line therapy in
`AML patients whoareineligible to receive standard induction therapy
`on the basis of high response rates and promising response durations
`in a Phase 1b/2 trial [27, 28]. These results have been very recently
`confirmed in the randomized, placebo-controlled, VIALE-A Phase 3
`trial which demonstrated the superiority of azacitidine plus venetoclax
`over azacitidine plus placebo in terms of CR rate (66.4% vs. 28.3%),
`duration of response (17.5 vs. 13.4 months), and OS (14.7 vs. 9.6
`months) [29]. These differences are highly statistically and clinically
`significant. This major breakthrough is reminiscent of the one made
`more than 40 years ago whenanthracyclines were added to cytarabine
`in patients eligible for intensive chemotherapy. Furthermore, the
`CR rate with azacitidine-venetoclax is >70% in the subgroups with
`IDH1 or IDH2 mutations [30]. These unprecedented results compared
`favorably with intensive chemotherapyin fit patients and generate
`hope for a cure in some patients. With the exception of the risk of
`tumorlysis syndrome and increased incidence of febrile neutropenia,
`no unexpected adverse event emerged, thus ensuring the widespread
`use of this combinationin this difficult-to-treat population of unfit or
`elderly patients. Venetoclax combined with low-dose cytarabine was
`also superior to low-dose cytarabine in the VIALE-C Phase3 trial [31].
`Thus, doublet azacitidine-venetoclax is becoming the new standard
`of care in patients ineligible for intensive induction chemotherapy and
`the standard arm to overcomein clinical trials. Planned or on-going
`clinical trials are already comparing this doubletto triplets with small
`molecule inhibitors or monoclonal antibodies. For example, the first
`results of the azacitidine-venetoclax-ivosidenib triplet may reach a
`100% rate of CR in treatment-naive AML patients with |DH1 mutations
`(EHALibrary. DiNardo C. 06/12/20; 294963; $143).
`There is an on-going debate on howto define patients unfit for
`intensive induction chemotherapy [32-34]. There are no absolute
`clear-cut criteria to unambiguously select the patients, and certain
`parameters used in the majority studies, such as performancestatus(a
`highly subjective criterion) or age, are questionable in this setting [34].
`It has been suggesting that comorbidity, physical capacities, and nutri-
`tional status may be more relevant than performancestatus or age [35].
`What about a 70-year-old patient with a normal karyotype, an ECOG
`performancestatus of 2 and no comorbidity? In routine, this patientis
`fit for chemotherapy especially when novaluable alternative is avail-
`able [36]. However, patients of the VIALE-A study could be included in
`the trial on the sole basis of the ECOGcriteria of 2; thereforeitis likely
`onco-hematology laboratories increase their turnaround in order to
`
`that this Phase3trial has included asubstantial numberof patients that allow clinicians to prescribe these agents in a timely manner. It has long
`were otherwisefit for chemotherapy outside clinical trials. Therefore,
`been postulated that AML represents an oncologic emergency and
`many physicianswill be tempted to replace intensive chemotherapy by
`should be treated withoutdelay, especially in younger patients treated
`azacitidine-venetoclax in older patients since CR rate and survival are
`by intensive chemotherapy [45]. However, recent studies have proven
`
`IDH AND FLT3 INHIBITORS, THE SMART
`3 |
`DRUGS WITH COMPANION BIOMARKERS
`
`With the developmentand recent approval of drugs targeting specific
`mutations such as FLT3, IDH1, and IDH2, it has become critical that
`
`

`

`6|WILEY
`
`RECHER
`
`that waiting a short period of time so as to characterise molecular
`alterations and design tailored treatments at diagnosis is safe [46, 47].
`It is now crucial that clinicians be knowledgeable on a panel of gene
`mutationsincluding at least FLT3, |IDH1, |IDH2, NPM1, CEBPA, and TP53
`at the time of diagnosis (48-72 h) in order to guide initial treatment.
`Furthermore, since AML clones and subclones are subject to clonal
`evolution under the therapeutic pressure of either chemotherapy
`or targeted agents, repeating molecular testing in each phase of the
`disease (refractory or relapse) should be mandatory. Recent advances
`inthe treatment of AML with FLT3 or IDHinhibitorsillustrate well this
`
`recent development.
`
`3.1 |
`
`IDH1 inhibitors
`
`Somatic mutationsofisocitrate dehydrogenase 1 (IDH1®132) genes are
`found in 6%-10% of AML patients [48]. |IDH1 mutations are mostfre-
`quent in AML with normal karyotype and associated with NPM1 and
`DNMT3A mutationsat diagnosis [49]. Their prognostic impact mainly
`dependson the mutational context [50-52]. Furthermore, IDH1 muta-
`tions, which have been described in clonal hematopoiesis, are consid-
`ered as early event during leukemogenesis, stable at relapse and thus,
`have emerged as promising therapeutic targets. It is also noteworthy
`that the molecular landscape of AML with IDH1 mutations observed at
`R/R disease under chemotherapyselection pressure differs from diag-
`nosis with an increased frequency of SRSF2, ASXL1, RUNX1, NRAS, and
`TP53 co-occurring mutations [49, 53].
`lvosidenib - an oral, targeted, small-molecule inhibitor of mutant
`IDH1 - has been evaluated as a single agent in a Phase 11 study in
`relapsed or refractory (R/R) AML with IDH1 mutation [53]. The fre-
`quencyof Grade 3 or higher treatment-related adverse events was low,
`mainly a prolongation of the QT interval, leukocytosis, and differen-
`tiation syndrome which are manageable [54]. CR or CR with partial
`hematologic recovery (CRh) was 30.4% with 21.8% CR, whereas CRi
`was 11.7%. It should be noted that clonal or subclonal mIDH1 had sim-
`
`ilar CR/CRh rates. Furthermore, mutation clearance was observed in
`
`21% of responding patients, thus demonstrating that deep response
`may be achieved in somepatients. The median duration of response
`was 9.3 months in CR patients. The median OS was8.8 months. Based
`on these promising results, ivosidenib was recently approved by the
`FDA.
`The mechanisms of resistance to ivosidenib were studied in
`
`patients whofailed to or relapsed after the response to this drug
`[55]. Receptor tyrosine kinase pathway mutations and mutations
`in NRAS and PTPN11 were significantly associated with the lack
`of
`response to ivosidenib.
`Interestingly, emerging mutations in
`patients who relapsed or progressed under ivosidenib were IDH or
`non-IDH-related. Indeed, mutations of resistance in a second site of
`
`The preliminary results of ivosidenib combined with azacitidine
`whentreating naive patients showed a CR rate of 70% and may reduce
`the emergence of mutantresistant clones [56].
`
`3.2 |
`
`IDH2 inhibitors
`
`Somatic mutations of the IDH2 gene, either IDH28149 or IDH2R172,
`occur in 5%-15% and 1%-4% of AML, respectively [48]. Like IDH1,
`IDH2 mutations are frequently found in normal karyotype AML [57,
`58]. At diagnosis, IDH2R14° mutations are associated with NPM1 and
`DNMT3A mutations whereasin the relapse/refractory setting, muta-
`tions of the SRSF2, DNMT3A, RUNX1, ASXL1, NRAS, and BCOR genes
`emerge as the most frequent co-mutations [11, 48, 59]. IDH2172 muta-
`tions are associated with DNMT3A and BCOR mutations and mutually
`exclusive with NPM1 and otherclass-defining mutations [49]. There-
`fore, AML with IDH28172 has been recognized as a defined subgroup
`of the AML genomicclassification[11].
`Enasidenib - an oral, targeted, small-molecule inhibitor of mutant
`IDH2 - has been evaluated as a single agent in a Phase 1 study in
`mutant IDH2 R/RAML patients and subsequently approved by the FDA
`[60]. A low frequency of Grade 3 or higher treatment-related adverse
`events was reported, mainly leukocytosis and differentiation syndrome
`[59, 61]. The overall response rate was 40.3% including 19.3% CR and
`6.8% CRi. The median OS was 9.3 months and reached 19.7 monthsin
`
`CR patients. CRs were observedin patients with subclonal |DH2 muta-
`tions, and a variantallele frequency of IDH2 mutant, which measures
`the mutational burden, was not associated with the response. Also,
`whereasin some CRpatients, |DH2 mutation clearance wasachieved,
`IDH2 mutational burden did not decrease in all responding patients
`during treatment, possibly due to the maturation of leukemic blasts
`into the functional neutrophils carrying the mutation. The mechanisms
`of resistance may involve the emergence of second-site IDH2 muta-
`tions, IDH2-mutant subclones with neomorphic mutationsin IDH1, co-
`occurring mutations in NRAS, and other MAPK pathwayeffectors or
`complex clonal evolution [59, 62, 63].
`A recent randomized Phase 2 trial of azacitidine versus azacitidine
`
`plus enasidenib in newly diagnosed AML patients unfit for intensive
`chemotherapy showeda significantly higher CR rate with the combina-
`tion comparedto azacitidine alone (53% vs. 12%) and a median dura-
`tion of response of 24.1 months in the combination arm (EHALibrary.
`DiNardo C. 06/12/20; 294959; $139).
`Whereas IDH inhibitors in combination with azacitidine yielded
`somevery interesting response rates compared to azacitidine alone,
`one openissue will be to determine which induction regimen to choose
`between azacitidine-venetoclax and azacitidine-IDH inhibitors. Will it
`
`be more appropriate to start with the standard azacitidine-venetoclax
`and reserve the IDH inhibitors for relapse? Or will triplets eradicate
`IDH1 or the emergence of IDH2®44° clones were detected in ~25%
`the diseaseatfirst line? Which inhibitor will we use in patients with
`other concomitant targetable mutations, such as FLT3 or TP53? In
`of resistant patients, whereas potentially actionable mutations in
`fit patients, the addition of ivosidenib or enasidenib to intensive
`genes such as FLT3, NRAS, or KRAS were also identified, thus indi-
`
`cating that molecular rescreening is important at each stage of the chemotherapyis under evaluation in an international Phase3trial
`disease.
`involving several cooperative AML study groups (NCT03839771).
`
`

`

`RECHER
`
`Several others IDH1 or IDH2 inhibitors including drugs that target
`both mutations are underinvestigation [64].
`
`3.3.
`
`|
`
`FLT3 inhibitors
`
`Mutations in the FLT3 gene are among the most common mutations
`in AML, occurring in up to 30% of patients [65]. There are twodis-
`tinct activating FLT3 mutations: internal tandem duplications (ITD)
`in the juxta-membrane domain and point mutations in the tyrosine
`kinase domain 2 (TKD). FLT3 mutations are associated with an aggres-
`sive disease course, especially FLT3-ITD which generally predicts an
`early relapse and poor prognosis. Through clonal selection under
`chemotherapy,a higher mutantallelic burdenis frequently observed at
`relapse, thus indicating that AML cells have become more addicted to
`FLT3 signalling. This is an important point because at least in preclin-
`ical settings, FLT3-mutantallelic burden andclinical status(i.e., diag-
`nosis versus relapse samples) are predictive of the response to FLT3
`inhibitors in AML [66].
`Midostaurin - a staurosporine analog with a multikinase inhibitory
`activity against KIT, PDGFR, PKC, VEGF, and FLT3 (amongst others)
`- wasthefirst FLT3 inhibitor to be approved for frontline therapy in
`AML patients with FLT3 mutationsandfit for intensive chemotherapy
`[67]. In the randomized Phase 3 trial RATIFY, midostaurin and placebo
`were added to standard “7+3”induction chemotherapy and high-dose
`cytarabine consolidation followed by a 12-monthssingle agent main-
`tenance in younger patients (18-60 years) with FLT3-ITD or FLT3-
`TKD mutations. The CR rates were similar in both groups. However,
`midostaurin significantly improved the 4-year overall OS from 44.3%
`to 51.4%, compared with placebo [7]. The benefit of midostaurin was
`observed in FLT3-ITD patients, whatever theallelic ratio, and in FLT3-
`TKDpatients. A subsequent exploratory analysis based on the FLT3-
`ITD patient of the RATIFY trial showed that the impact of midostau-
`rin wassignificant in the three prognostic subgroups of the ELN2017
`classification which includes NPM1, RUNX1, ASXL1, and TP53 as well
`
`as cytogenetic risk [68]. Furthermore, the benefit of midostaurin was
`independentfrom the allogeneic stem cell transplantation. A Phase 2
`study suggested that older patients aged 60-70 years mayalso benefit
`from midostaurin [69]. Midostaurin was approvedin Europefor induc-
`tion, consolidation, and maintenance, whereas it was only approved
`for induction and consolidation in the US indicating that the need
`for midostaurin as maintenanceis uncertain. However, this postulate
`could also apply to the consolidation phase since RATIFY was not
`designed to demonstrate that midostaurin treatment beyond induction
`chemotherapywasessential. Anyhow,since this approval and because
`FLT3 mutated patients present with a high tumor burdenat diagnosis,
`the results of FLT3 mutational screen must been foundrapidly.
`Two randomized Phase 3 trials with second generation FLT3
`inhibitors were recently conducted in R/R AML patients with FLT3
`mutations(gilteritinib) or FLT3-ITD only mutations (quizartinib) [70,
`71]. In both studies, the FLT3 inhibitor as a single agent was superior
`to the standard of care with high or low intensity chemotherapyin
`terms of response and OS. However, while gilteritinib has been broadly
`
`WILEY+
`
`approved in North America, Europe and Japan, quizartinib was only
`registered in Japan.
`Gilteritinib is an oral, small molecule inhibitor, highly selective of
`FLT3 acting against both FLT3-ITD and FLT3-TKD mutations and only
`acting marginally against cKIT [72, 73] which distinguishes it from
`quizartinib andlikely explains the weak myelosuppression observed in
`patients. Gilteritinib also targets AXL, another tyrosine kinase impli-
`cated in the resistance to chemotherapy and FLT3inhibitors [74, 75].
`In the pivotal Phase 3 ADMIRAL study, AML patients with R/R FLT3-
`mutated AML were randomized between 120-mg/daygilteritinib and a
`standard of care with high or low intensity regimen defined by physi-
`cians prior to randomization [70]. It is important to keep in mind that
`few patients of the ADMIRALtrial had previously been exposed to
`midostaurin, which is no longer the case. Gilteritinib induced higher
`CR/CRh and CR rates (34.0% vs. 15.3% and 21.1% vs. 10.5%, respec-
`tively) and significantly improved OS (median OS, 9.3 vs. 5.6 months).
`Adverse events were more frequent in the standard chemotherapy
`arm, with the exception ofliver transaminase elevations. QTc prolon-
`gation, differentiation syndrome, and lipase elevation are very rare
`events in the context of gilteritinib treatment (<5%), whereas pos-
`terior reversible encephalopathy syndromes have been exceptionally
`reported [54, 76]. Off-target activating mutations in genes of the
`RAS/MAPKpathway have been identified as a key mechanism of the
`resistance to gilteritinib and confirmed in patients of the ADMIRAL
`trial who relapsed ongilteritinib treatment in whom in-target FLT3-
`F691L mutations werealso detected [77, 78].
`Phase 3 randomizedtrials comparingfirst line intensive chemother-
`apy plus midostaurin or second generation inhibitors are ongoing,
`and many combinations with hypomethylating agents or targeted
`agents are also being investigated [79]. Furthermore, other novel FLT3
`inhibitors are under development, andit is foreseeable that clinicians
`may have a handful of FLT3 inhibitors in order to deal with clonal evo-
`lution, drug-drug interactions, adverse events, or co-morbid conditions
`just like BCR-ABL inhibitors for Philadelphia-chromosome-positive
`leukemia [80-83].
`Patients with FLT3-ITD mutations are generally candidatesto allo-
`geneic stem cell transplantation, even though the post-transplant
`relapse rate remains a problem [84, 85]. Interestingly, sorafenib - a
`multikinase inhibitor with potent activity against FLT3 - has demon-
`strated clinical activity in FLT3-ITD patients having relapsed after
`transplantation [86]. A subsequent comprehensive preclinical study
`elegantly demonstrated that sorafenib,
`like other FLT3 inhibitors,
`increased the IL-15 production by FLT3-ITD leukemic cells leading to
`the potentiation of the allogeneic CD8+ T cell response as well as
`disease eradication in preclinical models [87]. As a clinical translation
`of this finding, the randomized Phase 3 SORMAIN trial demonstrated
`that sorafenib maintenance therapy reducestherisk of relapse and
`death after transplantation in AML patients with FLT3-ITD mutations
`[88]. Reducing therisk of relapse after allogeneic stem cell transplanta-
`tion by maintenance therapy with non-cytotoxic drugsis an activefield
`of research going beyond FLT3inhibitors, and virtually all the small
`molecules inhibitors having shownefficacy in AML will be assessed in
`this context [89, 90].
`
`

`

`7| WILEY
`
`RECHER
`
`4 | TP53: WANTED, DEAD, OR ALIVE
`
`Although the TP53 tumor suppressor gene is the most frequently
`mutated gene in human cancer,its incidence in AML is relatively low
`(5%-20%) and increases with age or in therapy-related AML [48].
`Patients with TP53 mutations have one of the worst prognosis in AML
`because their disease is both chemo and immuneresistant, as shown
`
`by the poorresponserate to standard treatments including intensive
`chemotherapy and hypomethylating agents and the high rate of relapse
`after allogeneic stem cell transplantation [91-94]. Treatment of this
`AML subgroup is an urgent unmet medical need, and therefore, huge
`efforts have been made to drug the undruggable. Thefirst glimmer of
`hope came from eprenetapopt (APR-246), which is the first-in-class
`small molecule that selectively targets TP53 mutated cancercells [95,
`96]. Furthermore, eprenetapoptacts in synergy with azacitidine [97].
`In two recent phase2 trials designed for TP53 mutated myelodysplas-
`tic syndrome or AML, eprenetapopt combined with azacitidine induced
`an overall response rate of 62%-71% (44%-47% CR) with a median
`duration of response at 8-10.4 months. [98, 99]. Neurologic toxicity
`emerged as the main adverseeffect with this drug. Furthermore, it has
`been recently shown that AML with TP53 mutations are associated
`with an infiltration of cytotoxic lymphocytes in the tumor microenvi-
`ronment, indicating that immune intervention could be of valuein this
`subgroup of patients [100, 101]. Altogether and even though these
`data are immature, there is reasonable hope to improve the outcome
`of patients with TP53 mutated-AML in the near future.
`
`5
`
`| CHEMOTHERAPY’S NOT DEAD(YET)
`
`A sizable proportion of intermediate and good-risk AML patients are
`cured by intensive chemotherapyin one shot(i.e., without the need
`for allogeneic stem cell transplantation and withoutrelapse). For these
`patients who generally receive one 7 + 3 induction cycle then three
`cycles of intermediate-to-high dose cytarabine, treatmentis definitely
`completed in 6 months, and thereafter, the patients are treatment-
`free for the rest of their life. In these patients, the added value of new
`drugswill be challenging to demonstrate both in termsof efficacy and
`duration of treatment because most new drugs have been developed
`so as to be administered on a long-term basis. However, well-known
`adverse eventsincluding profound myelosuppression,gastro-intestinal
`toxicity, severe mucositis, and infections as well as the strong impact
`of chemotherapyon quality oflife remain of concern. CPX-351,a dual-
`drugliposomal combination of daunorubicin and cytarabine with a syn-
`ergistic drug ratio, was approvedfor the treatmentof adults with newly
`diagnosed therapy-related AML or AML with myelodysplasia-related
`changes[102]. Long-term results of the pivotal phase 3 trial shown at
`ASH 2020 confirmed the superiority of CPX-351 over standard “3+7”
`chemotherapy and the particular good outcomeof patients who were
`allografted in first response after CPX-351 [103]. Interestingly, CPX-
`351 accumulates in the bone marrow where it has been shown to
`
`be taken up to a greater extent by AML cells than normal bone mar-
`row cells and sparing normal tissues [104]. In a clinical context, this
`
`translates into improvedefficacy (a higher responserate) but also into
`increased tolerability to induction chemotherapy. Nurses were proba-
`bly amongthefirst to notice this curious effect of CPX-351 compared
`to free daunorubicin and cytarabine: much less mucositis, gastroin-
`testinal (Gl) toxicity, and nohairloss.It is generally recognized by care-
`givers that CPX-351 is better tolerated than the standard 7 + 3, even
`though the Phase3 trial did not clearly demonstratethis point. Further-
`more, when ranking adverse events, it is noteworthy that hair-loss is
`the most commonside effect reported as severe by the patients, while
`caregivers are more proneto declareinfections, suggesting that quality
`of life with CPX-351 should be better than with 7+3 [105]. Therefore,
`it is tempting to foresee that CPX-351 indications could extend beyond
`the label to be broadly applied in de novo AML.In this regard, the results
`of the ongoing Phase 3 trial of the German AMLSGstudy group, cur-
`rently assessing CPX-351 versus standard “7+3” in newly diagnosed
`AML andintermediate- or adverse genetics (>18 y) (NCT03897127),
`will be of great importance for AML patientsfit for chemotherapy.
`
`6 | REVISITING THE CONCEPT OF REMISSION
`MAINTENANCE WITH ORAL THERAPY IN AML
`
`Preventing relapse after induction and consolidation therapy in
`AML has long been a major challenge especially in older patients
`ineligible for allogeneic stem cell
`transplantation [106]. While
`there have been some interesting data with drugs such as the
`histamine dihydroc

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket